Development of Dual Drug Eluting Cardiovascular Stent with Ultrathin

influences the performance of the multi-drug eluting stents. .... vivo study, the commercial stents and bare metal stents were gifted by an Indian ste...
0 downloads 0 Views 9MB Size
Subscriber access provided by CAL STATE UNIV BAKERSFIELD

Controlled Release and Delivery Systems

Development of dual drug eluting cardiovascular stent with ultrathin flexible PLCL coating Purandhi Roopmani, Santhosh Satheesh, David C Raj, and Uma Maheswari Krishnan ACS Biomater. Sci. Eng., Just Accepted Manuscript • DOI: 10.1021/acsbiomaterials.9b00303 • Publication Date (Web): 10 Apr 2019 Downloaded from http://pubs.acs.org on April 16, 2019

Just Accepted “Just Accepted” manuscripts have been peer-reviewed and accepted for publication. They are posted online prior to technical editing, formatting for publication and author proofing. The American Chemical Society provides “Just Accepted” as a service to the research community to expedite the dissemination of scientific material as soon as possible after acceptance. “Just Accepted” manuscripts appear in full in PDF format accompanied by an HTML abstract. “Just Accepted” manuscripts have been fully peer reviewed, but should not be considered the official version of record. They are citable by the Digital Object Identifier (DOI®). “Just Accepted” is an optional service offered to authors. Therefore, the “Just Accepted” Web site may not include all articles that will be published in the journal. After a manuscript is technically edited and formatted, it will be removed from the “Just Accepted” Web site and published as an ASAP article. Note that technical editing may introduce minor changes to the manuscript text and/or graphics which could affect content, and all legal disclaimers and ethical guidelines that apply to the journal pertain. ACS cannot be held responsible for errors or consequences arising from the use of information contained in these “Just Accepted” manuscripts.

is published by the American Chemical Society. 1155 Sixteenth Street N.W., Washington, DC 20036 Published by American Chemical Society. Copyright © American Chemical Society. However, no copyright claim is made to original U.S. Government works, or works produced by employees of any Commonwealth realm Crown government in the course of their duties.

Page 1 of 53 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

ACS Biomaterials Science & Engineering

Development of dual drug eluting cardiovascular stent with ultrathin flexible PLCL coating

Purandhi Roopmani1,2, Santhosh Satheesh4, David C. Raj2, Uma Maheswari Krishnan1,2,3* 1 2 3 4

Centre for Nanotechnology & Advanced Biomaterials (CeNTAB)

School of Chemical & Biotechnology, SASTRA Deemed University, Thanjavur, Tamilnadu

School of Arts, Science & Humanities, SASTRA Deemed University, Thanjavur, Tamilnadu

Jawaharlal Institute of Post Graduate Medical Education and Research (JIPMER), Department of Cardiology, Pondicherry -605 006, India

____________________________________________________________________ *Corresponding Author Prof. Uma Maheswari Krishnan Ph. D. Dean, School of Arts, Science & Humanities (SASH) Professor, School of Chemical & Biotechnology SASTRA Deemed University, Thanjavur – 613 401 Tamil Nadu, India. Ph.: (+91) 4362 264101 Ext: 3677 Fax: (+91) 4362 264120



1

ACS Paragon Plus Environment

ACS Biomaterials Science & Engineering 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

E–mail: [email protected]



2

ACS Paragon Plus Environment

Page 2 of 53

Page 3 of 53 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

ACS Biomaterials Science & Engineering

ABSTRACT The pleiotropic effects of atorvastatin-fenofibrate combination can be effectively harnessed for sitespecific therapy to minimize stent-related complications. The present study aims to utilize the pleiotropic effects of these two drugs entrapped in a uniform and defect-free coating of poly(Llactide-co-caprolactone) (PLCL) on a stainless steel stent to overcome stent-associated limitations. The stent coating parameters were optimized using ultrasonic spray coating technique to achieve a thin, smooth and defect-free dual drug-loaded polymer coating on the stent. The dual drug-loaded polymer coated stent was characterized for surface morphology, thickness and coating integrity. In vitro drug release kinetics of the fabricated stent reveals a sustained release of both drugs for more than 60 days. Significant reduction of thrombus formation and adhesion of LPS-stimulated macrophages on the dual drug containing polymer-coated stent indicates that the drug combination possesses anti-thrombotic and anti-inflammatory effects. The combination did not adversely influence endothelialization but significantly retarded smooth muscle cell proliferation indicating its potential to overcome restenosis. No bacterial biofilm formation was observed on the stent due to the anti-bacterial activity of atorvastatin. A rat subcutaneous model was used to evaluate the biocompatibility of the coated stent and compared with the commercial stent. MicroCT, SEM and morphometric analyses revealed that the coated stents exhibited excellent histocompatibility with no inflammatory response as evidenced from the cytokine levels measured 28 days post-implantation. Our data demonstrates for the first time that the combination of atorvastatin and fenofibrate can be successfully employed in cardiovascular stents to overcome the current limitations of conventional drug-eluting stents.



3

ACS Paragon Plus Environment

ACS Biomaterials Science & Engineering 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Keywords: Dual drug-eluting stent, ultrasonic spray technique, in vivo biocompatibility, Atorvastatin-fenofibrate

1. INTRODUCTION Drastic changes in lifestyle, dietary habits and environment quality in the modern era has resulted in increased incidence of atherosclerosis across the globe.1 The extensively employed therapeutic intervention for this condition is the deployment of stents at the blocked site to restore circulation through the blood vessels.2 Regardless of the success and advancement in stent technology, recurrence of restenosis and late stent thrombosis continue to limit the performance of coronary interventions. Restenosis arises due to imbalance in the proliferation rates of endothelial and smooth muscle cells whereas thrombosis occurs due to adhesion and activation of the platelets by the stent surface.3, 4 Conventionally, drug-eluting stents containing anti-proliferative drugs such as sirolimus, everolimus, paclitaxel, etc., have been employed in combination with antiplatelet agents such as clopidogrel, aspirin, dabigatran, rivaroxaban etc.5 The past generations of drug-eluting stents have clinically proven their efficiency in reducing the in-stent restenosis by 20%.6 However, though the anti-proliferative drugs retard the migration and proliferation of human vascular smooth muscle cells, they also lead to poor endothelialization thereby initiating the pro-thrombotic cascade and escalation of inflammatory responses.7 In addition, long-term anti-thrombotic therapy causes several adverse effects, especially bleeding risks, thereby lowering the quality of life.8 To mitigate stentassociated complications, new drugs, nanotextured stent surfaces, bioresorbable platform and other surface functionalization techniques have been explored.9 However, such expensive and complex strategies produced less satisfactory results in clinical trials. Further, epidemiological variations in the therapeutic outcomes have also been reported.10



4

ACS Paragon Plus Environment

Page 4 of 53

Page 5 of 53 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

ACS Biomaterials Science & Engineering

Due to the shortcomings of monotherapy employed in conventional drug-eluting stents, combinations of drugs have been explored as an alternative. Blends of anti-proliferative drugs with anti-thrombotic or re-endothelialization promoter 11 have been explored for use in multi-drug eluting stents. The choice of therapeutic agents and their release kinetics from the coating employed influences the performance of the multi-drug eluting stents. Many combinations such as r-PEGhirudin and iloprost, cilostazol,

15

12

paclitaxel and pimecrolimus,

sirolimus and paclitaxel

16

13

sirolimus and estradiol,

14

paclitaxel and

etc., have been attempted but had low success in clinical

trials. The clinical trials revealed that there was no significant reduction in the thrombosis events using the above combinations. 11 The fast release of drugs from the stent could have been a probable reason for the lack of significant anti-thrombotic activity. Several other combinations are also being investigated but clinical trial data is not yet available.11 Moreover, the lack of specificity manifested through poor wound healing at the stented site is another drawback of typical anti-proliferative drugs used in coronary stents.17 Therefore, it is evident that an ideal combination of multi-action drugs should possess specific inhibitory action towards vascular smooth muscle cells apart from exhibiting endothelial cell-protective, anti-inflammatory and anti-thrombotic effects. In this context, the present work explores the combination of two well-known lipid-lowering drugs atorvastatin and fenofibrate that have been reported to exert beneficial effects on cardiovascular alignment through non-lipid lowering mechanisms18 but have never been studied earlier as a combination in coronary stents.

Atorvastatin, a HMG-CoA (hydroxymethyl glutaryl-coenzyme A) reductase inhibitor, has been shown to inhibit the activation and invasion of vascular smooth muscle cells by regulating various intracellular signaling pathways such as NF B (nuclear factor



5

ACS Paragon Plus Environment

-B), 19 RhoA-Rho related kinase, 20,

ACS Biomaterials Science & Engineering 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

21

and other small GTPase family proteins.22 Moreover, it has been found to exhibit a stabilizing

effect against vascular injury caused by chemo-attractants like platelet-derived growth factor-BB.23, 24

Other important pleiotropic effects of atorvastatin include its ability to elevate eNOS (endothelial

nitric oxide synthase) and expression levels of endothelial junction proteins,25, 26 escalate nitric oxide production and regulate Akt/PKB (Protein kinase B) signaling27 in endothelial cells. Atorvastatin has also been shown to retard atherosclerosis-induced release of pro-inflammatory cytokines.28, 29 These pleiotropic properties suggest that atorvastatin can counter endothelial dysfunction.

Fenofibrate, a peroxisome proliferator-activated receptor-α (PPAR-α) activator, exerts antiinflammatory effect against vascular smooth muscle cell induced interleukins.30 This property may be beneficial in lowering the risk of implant-based inflammation. An earlier report has described the inhibition of VSMC synthesis stimulated via platelet-derived growth factor (PDGF) by treatment with fenofibrate.

31

Fenofibric acid, an active form of fenofibrate has been found to retard the

activity of PDGF independent of cholesterol mechanism. Also, fenofibrate exhibits an antiatherogenic effect on vascular smooth muscle cells by decreasing the biosynthesis of proteoglycans. 32

Further, it has been demonstrated to significantly decrease arterial thrombus formation and

platelet aggregation in animal models by inhibiting thromboxane A2 receptor and cyclooxygenase1.33 Fenofibrate also prevents endothelial dysfunction by increasing antioxidant-mediated production of the vasodilator nitric oxide.34 Though both drug molecules have independently and in combination displayed cardio-protective properties, they have never been considered as a possible combination for stent-based delivery. The present work for the first time aims to fabricate, optimize and characterize a polymer coating containing the two lipid-lowering drugs over a metallic stent and evaluate its in vivo biocompatibility. The biodegradable polyester poly(L-lactide-co-caprolactone)



6

ACS Paragon Plus Environment

Page 6 of 53

Page 7 of 53 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

ACS Biomaterials Science & Engineering

(PLCL) (70:30) was chosen as the polymer matrix over the commonly employed PLGA (poly(lactide-co-glycolide). This is because PLGA coatings are more prone to brittle fracture depending on their lactide content that may lead to complications during crimping and stent deployment.35 PLCL is biocompatible polyester that has been employed for tissue engineering applications and exhibits controlled degradation and elastic nature, 36 both of which are valuable for stent applications.

2. MATERIALS AND METHODS 2.1 Materials. Poly(L-lactide-co-ε-caprolactone) (7030LCL MW=230kDa) was purchased from Evonik Degussa Corporation (Birmingham, USA). Atorvastatin calcium (ATS) (MW = 1209.4 g/mol) was obtained from Swapnroop Drugs & Pharmaceuticals (India) and Fenofibrate (FF) (F6020 MW = 360.83 g/mol) was purchased from Sigma-Aldrich (USA). Acetone of HPLC grade from Merck, India was used to dissolve the polymer and drugs. All organic solvents and chemicals used were of analytical grade. All metal stents (316L stainless steel, 7 mm in length) and stent delivery catheters were provided by JIPMER (Pondicherry, India) for optimizing the stent coating. For in vivo study, the commercial stents and bare metal stents were gifted by an Indian stent manufacturer. Multi-analyte ELISA kit was purchased from Qiagen, USA.

2.2 Fabrication of dual drug loaded PLCL stents. MediCoat (DES1000, Sono-Tek Corporation, Milton, NY), a customized ultrasonic spray system was used for coating the bare metal stent. The stainless steel stents (length 7 mm, diameter 3 mm) were cleaned with acetone and distilled water, air-dried and mounted on the mandrel of 3 mm diameter. For spray coating, 3% of PLCL and drugs (1:1 ratio) were dissolved in HPLC grade acetone and loaded in a syringe pump. The drug ratio was chosen based on our earlier in vitro studies using human umbilical vein endothelial cells (HUVECs)



7

ACS Paragon Plus Environment

ACS Biomaterials Science & Engineering 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

and human vascular smooth muscle cell (hVSMCs). The flow rate, ultrasonic power, gas pressure, distance between nozzle and stent, rotation speed, and other coating parameters were thoroughly examined for obtaining a defect-free smooth coating. After coating, stents were vacuum dried for 2 days. Surface morphology of the coated stents was observed under field emission scanning electron microscopy (FE-SEM, JSM 6701F, JEOL, Japan).

2.3 Balloon expansion test. The quality and integrity of therapeutic coating were evaluated using balloon expansion test. Dual drug coated stents were manually compressed on a balloon catheter up to a diameter of 1 mm. After imaging, crimped stents were then expanded to 3 mm diameter by applying a pressure of 12 psi. Morphology of coating after the expansion was examined by using FE-SEM (JSM 6701F, JEOL, Japan).

2.4 In vitro drug release study. Drug release studies on the dual drug-loaded PLCL stents (n=3) were performed in PBS (phosphate buffered saline) medium (pH 7.4) at 80 rpm for 80 days at 37oC. The medium was collected and replaced with fresh medium at every time point. The collected medium was analyzed using UV-Visible spectrophotometry (Lambda 25, Perkin Elmer, USA) at 246 nm and 286 nm for atorvastatin and fenofibrate respectively. To observe the morphological changes in the coating after 80 days of drug release, the surface of the stents was imaged using SEM (TESCAN Vega, Czech Republic).

2.5 In vitro degradation study. Dual drug-loaded PLCL coated stents were incubated in PBS (pH 7.4) at 37oC under constant stirring at 80-120 rpm. The medium was replaced with fresh medium every alternate day. At specific time points, the stents were removed, rinsed with distilled water, air-



8

ACS Paragon Plus Environment

Page 8 of 53

Page 9 of 53 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

ACS Biomaterials Science & Engineering

dried and subjected to vacuum drying. The change in surface morphology of the stent coated with dual drug loaded polymer was imaged using SEM.

2.6 Cell adhesion study and immunostaining. Human umbilical vein endothelial cells (HUVECs) (Hi-Media, India) were cultured in endothelial growth medium-2 EGM-2 (Lonza, USA) and human vascular smooth muscle cells (hVSMC) were maintained in Dulbecco’s modified Eagle’s medium (DMEM) (Gibco, USA) supplemented with 10% fetal bovine serum, 1% penicillin-streptomycin at 37oC and incubated in 5% CO2 (NU-8500, Nuaire, USA). For the study, bare stainless steel stents and dual drug-loaded PLCL coated stents were UV sterilized and incubated with 50,000 cells per well. At day 1 and day 3, the morphology of adhered HUVEC and hVSMC was observed using SEM after fixing the cells using 2.5 % glutaraldehyde. For immunostaining, the adhered cells were fixed using paraformaldehyde (4%) for 10 min followed by TritonX-100 (0.3%) in PBS buffer for 10 min at room temperature. The samples were then incubated for 1 h using blocking solution (1 % bovine serum albumin). Post-incubation, samples were washed with DPBS followed by overnight incubation with antibodies against endothelial cell adhesion molecule (CD31) (Abcam, USA) and αSMA (alpha-smooth muscle cell actin) (1:100) (Abcam, USA) at 4oC. This was followed by removal of primary antibody. The samples were washed with DPBS and incubated with goat anti-mouse IgG (H+L) Alexa Fluor 555 (Invitrogen, USA) for 1 h. The samples were then observed using fluorescence microscopy (Nikon Eclipse, TS100, Japan).

2.7 Whole blood adhesion and APTT test. The dual drug loaded PLCL stent was further tested for in vitro hemocompatibility using whole blood. Bare stainless steel stent, PLCL coated stent and dual drug-loaded PLCL coated stent were incubated with freshly collected human blood for 1 h at 37oC



9

ACS Paragon Plus Environment

ACS Biomaterials Science & Engineering 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 10 of 53

under static conditions. After incubation, the stents were washed gently with phosphate buffered saline and the adhered cells were fixed by overnight incubation with 2.5% glutaraldehyde followed by dehydration of the stents by sequential treatment using 50%, 70%, 90% and 100% ethanol. After fixing, the stents were vacuum dried and imaged using FE-SEM. To evaluate the in vitro anticoagulation property, APTT (activated partial thromboplastin time) test was performed using blood plasma. Bare metal, PLCL coated and dual drug-loaded PLCL coated stents (n=3) were incubated with100 μL of freshly isolated platelet poor plasma for 15 min at 37oC. After incubation, 100 μL of cephaloplastin reagent was added followed by CaCl2 solution (0.025 M). The sample was left undisturbed for 3 min. The time taken for the appearance of a fine thread of clot was measured using coagulometer (STA Compact Stago, USA). The results were compared with those obtained for platelet poor plasma.

2.8 In vitro inflammatory response. Stent samples were cultured with IC-21 mouse macrophage cell line (NCCS, Pune) stimulated with 100 ng/mL of lipopolysaccharide (LPS) (Sigma Aldrich, USA) to test their anti-inflammatory response. Cells were maintained in RPMI medium (Invitrogen, USA) with 10% FBS and 1% penicillin-streptomycin (Gibco, USA). LPS-stimulated IC-21 cells with the seeding density of 50,000 cells per well were incubated with bare stainless steel, PLCL coated or dual drug-loaded PLCL coated stents for 24 h. The cell morphology was observed under phase contrast microscopy (Zeiss Axio Vert A1, USA). Further, the cells adhered to the surface of the stents were fixed with 2.5% glutaraldehyde and imaged using SEM.

2.9 Biofilm formation. Sterilized stents were incubated with tryptic soy broth (TSB) (Hi-Media, India) inoculated with 2% bacterial culture of Staphylococcus aureus (ATCC 25923). After 24 h of



10

ACS Paragon Plus Environment

Page 11 of 53 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

ACS Biomaterials Science & Engineering

incubation, stents were washed with PBS and the adhered bacteria were fixed with 2.5% glutaraldehyde and observed using scanning electron microscopy.

2.10 In vivo studies. In vivo biocompatibility of the dual drug-loaded PLCL stent was tested by subcutaneous implantation in Wistar rats. All animal experiments were carried out after approval of the study protocol from the Institutional Animal Ethics Committee (471/SASTRA/IAEC/RPP). Twenty-four male Wistar rats about 6 - 8 weeks old were chosen for the study. They were segregated and acclimatized before the surgery. Animals were anaesthetized using ketamine (80 mg/kg ip) (Aneket, Neon Laboratories, India) and xylazine (6 mg/kg ip) (Tamil Nadu Government supplies, India). The hair was removed from the dorsal side of the rats and the skin was sterilized with 70% ethanol. A small incision was made to expose the dorsum. A deep subcutaneous pouch was created using sterile surgical scissors. Ethylene oxide (ETO) sterilized stents were implanted in the pouch using 1 mL sterile syringe. Once the stent was secured in the subcutaneous pouch, the incision was closed with sterile absorbable surgical sutures (ETHICON, Johnson & Johnson, India) and povidone iodine was applied near the suture.

2.11 Micro-CT analysis. X-ray scan of the implanted site was taken on 7th and 28th days poststenting using Micro-CT SKYSCAN (Bruker, Germany). Animals with commercial and dual drugloaded PLCL coated stent were anaesthetized and prepared for the scan. Each animal was scanned for 180o and a three-dimensional visualization was obtained by enhancing the contrast using the Micro-CT software (CTVox).



11

ACS Paragon Plus Environment

ACS Biomaterials Science & Engineering 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 12 of 53

2.12 Necropsy. After 7 and 28 days of stent implantation, the animals were euthanized using CO2 inhalation method and the skin was cut open to expose the site of stent implantation. Images of the subcutaneous pocket with stent were taken. Stents along with surrounding tissue were isolated and carefully stored in 10% neutral buffered formalin solution for histopathological analysis. To analyze the tissue adhesion and stent coating quality, the samples were fixed with 2.5% glutaraldehyde and imaged using SEM (TESCAN Vega, Czech Republic).

2.13 Quantification of Inflammatory cytokines. Multi-Analyte ELISA kit (Qiagen, USA) was used to detect the levels of inflammatory markers in the serum collected from the animals before euthanasia. The kit quantifies the cytokines like interleukin 1A (IL-1A), interleukin 1B (IL-1B), interleukin 2 (IL-2), interleukin 4 (IL-4), interleukin 6 (IL-6), interleukin 8 (IL-8), interleukin 10 (IL-10), interleukin 12 (IL-12), interleukin 17A (IL-17A), and interferon γ (IFN-γ), tumor necrosis factor α (TNF-α), granulocyte macrophage-colony stimulating factor (GM-CSF) and Regulated on activation, normal T cells expressed and secreted (RANTES), which are involved in inflammation and T-cell biology. This assay was used to evaluate inflammation caused by the dual drug eluting stent and was compared with sham control. Following the manufacturer’s protocol, 50 μL of serum samples collected from animals 28th day post-stenting or standards were added to appropriate wells. The samples were incubated for 2 h at room temperature. After washing, 100 μL of detection antibody solution was added and incubated for 1 h. After incubation, the strips were washed 4 times using the washing buffer and 100 μL avidin-HRP solution was added and incubated at room temperature for 30 min. Finally, 100 μL of the development solution was added to each well and incubated in the dark for 15 minutes at room temperature. Subsequently, 100 μL of stop solution



12

ACS Paragon Plus Environment

Page 13 of 53 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

ACS Biomaterials Science & Engineering

was added to each well, and the absorbance of the samples was measured at a wavelength of 450 nm using a multimode reader (Infinite M200, Tecan, Austria).

2.14 Statistical analysis. The results of all experiments performed in triplicate were analyzed using one-way ANOVA. The statistical significance (* p < 0.05) was determined using student-T test.

3. RESULTS AND DISCUSSION 3.1 Stent coating. Irregularities in stent coating can cause serious complication of thrombosis within 24 h of stent implantation. Hence it is very important to develop a smooth and uniform therapeutic coating on the stents without any defects. FE-SEM images (Figure 1) of polymer coated stent show the effect of various coating parameters on surface morphology and uniformity of the coating. The complete coverage of the stent with smooth surface was observed at 3 wt% polymer solution which was selected for further trials. Similarly, 1.5 W of ultrasound frequency, 0.5 psi of gas pressure and 6 mm distance between nozzle and stent were optimized based on a uniform shape of the spray of the polymer solution and formation of a uniform coating on the surface with less defects. These conditions were kept constant while optimizing major stent coating parameters. Figure 1[A] shows the SEM images of the stent coated at various flow rates from 0.02 mL/min to 0.1 mL/min while the rotation speed, feeding speed and delay time was kept constant at 100 rev/min, 0.300 cm/s and 2 s respectively with the number of loops kept at 2. At a low flow rate of 0.02 mL/min, the spray was unable to cover the entire stent while at the high flow rate of 0.1 mL/min webbing defects were formed near the struts due to a large amount of polymer deposition. Moderate flow rates did not show defects but based on the smoothness of the coating, the flow rate of 0.06 mL/min was chosen as optimum. The rotation speed of the stent was varied from 70 rpm to 150 rpm for obtaining the



13

ACS Paragon Plus Environment

ACS Biomaterials Science & Engineering 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 14 of 53

defect-free coating, with the flow rate maintained at 0.06 mL/min, feeding speed at 0.300 cm/s, delay time at 2 s and number of loops at 2. Both low as well as high rotation speeds, resulted in webbing defects (Figure 1[B]). At 100 rpm and 120 rpm no defects were observed but depending on better surface uniformity, 100 rpm was chosen as optimum for a smooth coating. Feeding or translation speed that defines the movement of the stent from one position to another under the spray was varied from 0.150 cm/s to 0.350 cm/s at 0.06 mL/min flow rate, 100 rpm rotation speed, delay time of 2 s and 2 cycles. Webbing defects were noticed at a low speed of 0.150 cm/s due to excessive deposition of polymer, whereas at 0.200 cm/s uneven coating was observed. At a high feeding speed of 0.350 cm/s, the coated surface was rough because of the non-uniform evaporation of the solvent. Therefore 0.250 cm/s and 0.300 cm/s with minimum inhomogeneity and defects were chosen for further trials. The delay time between each loop of the stent was varied from 1 s to 8 s. Based on the uniform and smooth coating obtained, 6 s and 8 s were chosen for further trials. Finally, the number of loops was examined from 2 to 10 loops at a flow rate of 0.06 mL/min, rotation speed of 100 rpm, feeding speed of 0.300 cm/s and delay time of 2 s. For 5 number of loops, an even smooth coating with the thickness of 1 μm was obtained and was chosen as optimum.

Based on the initial optimization trials, two different sets of conditions with different feeding speed and delay time were shortlisted for single drug coating. The drug-loaded polymer coating was evaluated using FE-SEM. Results revealed that feeding speed of 0.300 cm/s, flow rate of 0.06 mL/min, rotation speed of 120 rpm, 5 loops and delay time of 8 s show no phase separation and formed a uniform coating due to steady evaporation of solvent and therefore was selected for dual drug coating (Supplementary information. Figure S1, FE-SEM images of drug loaded stents at different coating conditions).



14

ACS Paragon Plus Environment

Page 15 of 53 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

ACS Biomaterials Science & Engineering

[A]

0.02mL/min

[B]

70rpm

[C]

0.150cm/s

[D]

1s

[E]

2 loops

0.08mL/min

0.1mL/min

120rpm

150rpm

0.250cm/s

0.300cm/s

0.350cm/s

2s

4s

6s

8s

3 loops

5 loops

0.04mL/min

80rpm

0.06mL/min

100rpm

0.200cm/s

8 loops

10 loops

Figure 1. Scanning electron micrographs of the surface of PLCL coated stent depicting the effect of various conditions during optimization of different coating parameters [A] flow rate, [B] rotation speed, [C] translation speed, [D] delay time and [E] number of loops. Scale bar represents 100 μm. The arrows show the coating defects.

Figure 2 shows the FE-SEM images of the stent with smooth and wrinkle-free dual drug-loaded polymer coating without any coating defects. The optimized therapeutic coating was further characterized for its stability and tested for hemocompatibility before the in vivo biocompatibility study. The scratch test performed on the coating to measure the coating thickness revealed that the dual drug-loaded polymer coating thickness was 1.4 μm, which is comparatively thinner than the



15

ACS Paragon Plus Environment

ACS Biomaterials Science & Engineering 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 16 of 53

coatings in commercially available drug-eluting stents, which are about 5-6 μm thick.37 The thinner layer of the polymer will ensure the least obstruction to blood flow through the stented vessels.

[A]

[B]

[C]

[D]

Figure 2. FE-SEM of dual drug-loaded PLCL coated stent at [A] low magnification and [B] at high magnification using optimized coating parameters, [C] scratch test to measure the thickness of drug-loaded polymer coating and [D] Thickness of coating at high magnification with annotation.

Ultrasonic spray technique involves translation and rotational movements of the stent, which ensures uniform coverage in both planar and non-planar regions of the stent by the ultrafine mist of the polymer spray.

38

Thickness of the coating is in turn determined by the density of spray that is

optimized through controlling the feed rate of the gas and liquid in to the spray nozzle. A uniform droplet size spray increases the uniformity of the coating thickness. 39



16

ACS Paragon Plus Environment

Page 17 of 53 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

ACS Biomaterials Science & Engineering

3.2 Balloon expansion test. An important requirement of polymer coating over stents is their flexibility. The stents are generally crimped over the balloon and are then expanded during the deployment. The polymer coating should withstand these extreme changes in pressure and deformative stresses without undergoing any brittle fractures. The presence of drug molecules in the polymer matrix may also alter the flexibility of the polymer film. To characterize the flexibility of the dual drug-loaded PLCL coating, balloon expansion test was performed. It was found that the dual drug-loaded PLCL coating displayed no cracking or peeling after the crimping and expansion cycle (Figure 3) indicating that the presence of both drugs in PLCL film did not alter its adhesion or flexible nature. At higher magnification, a smooth and uniform coating was observed without any micro-defects or delamination suggesting that the drug-loaded polymer coating possesses sufficient elasticity that can remain stable even after the expansion stress. This finding augers well for the use of this dual drug-loaded polymer as a therapeutic coating on stents, which may undergo repeated deformation.40

[A]

[C]

[B]

[D]

Figure 3. FE-SEM images of dual drug-loaded PLCL coated stent [A] before balloon expansion (crimped position) and [B] after balloon expansion test. [C] and [D] High magnification images of the stent after expansion. Scale bar for C & D images represents 100 μm.



17

ACS Paragon Plus Environment

ACS Biomaterials Science & Engineering 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 18 of 53

3.3 In vitro release profile. The release profile of both drugs from the polymer coating was investigated at 37oC in PBS under the dynamic condition at 80 rpm and the results are presented in Figure 4. A sustained release was observed for both drugs over a period of 80 days. The cumulative release of atorvastatin was higher when compared to fenofibrate at all the time points. Fenofibrate is uncharged while atorvastatin is anionic and this difference may influence the distribution of the two drugs within the PLCL matrix. The atorvastatin may prefer to localize towards the periphery while the fenofibrate partitions into the interior. This accounts for the higher burst release observed in the case of atorvastatin. SEM images of this therapeutic coating on stents after 80 days reveal that the coating is still present on the stent but with a rough surface morphology (Figure 4). This indicates that the polymer film has degraded slowly. Mathematical modeling of the drug release kinetics over 80 days was performed and the regression coefficients were calculated from the standard equations (Table 1).

Table 1. Regression coefficients of various mathematical models of drug release at different time points Overall (0-80 Days)

0-72h

0-42 Days

42-80 Days

Models ATS

FF

ATS

FF

ATS

FF

ATS

FF

Zero order

0.6569

0.7150

NC

NC

NC

NC

0.6465

0.6400

First order

0.6733

0.7245

NC

NC

NC

NC

0.6679

0.6608

Higuchi

0.7441

0.7594

NC

NC

NC

NC

0.5271

0.5228

0.7503

0.7764

0.5702

0.6734

0.8910

0.9214

0.6552

0.6490

Hixson-Crowell

0.6696

0.7222

NC

NC

NC

NC

0.6677

0.6578

Hopfenberg

0.6733

0.7244

NC

NC

NC

NC

0.6685

0.6605

KorsmeyerPeppas



18

ACS Paragon Plus Environment

Page 19 of 53 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

ACS Biomaterials Science & Engineering

Baker-Lonsdale

0.7338

0.7512

NC

NC

NC

NC

0.5018

0.5057

Makoid-Banakar

0.8683

0.8798

0.7270

0.7754

0.9441

0.9539

0.9563

0.9524

Peppas-Sahlin

0.7709

0.7962

0.7814

0.8188

0.8923

0.9229

0.7400

0.7335

Quadratic

0.6739

0.7261

NC

NC

NC

NC

0.6718

0.660

Weibull

0.8636

0.8737

0.9634

0.9971

0.9350

0.9489

0.9177

0.9118

Logistic

0.7185

0.7556

0.5725

0.6745

0.8899

0.9205

0.6935

0.6737

Gompertz

0.6541

0.6932

0.5796

0.6813

0.8865

0.9166

0.7317

0.7088

Probit

0.6896

0.7258

0.5760

0.6782

0.8882

0.9185

0.7046

0.6868

NC (not correlated)

It is observed that the release profiles of both atorvastatin and fenofibrate did not agree with most of the kinetic models over the total 80-day period. The closest agreement was with the Weibull model that describes the release from a matrix-based system. This suggests that the drug release is controlled by different parameters at different time points. When the drug release profiles were modeled for specific time points, the initial release up to 72 h was found to agree with the Weibull kinetics for both drugs while at later time points, the Makoid-Banakar model and Weibull kinetics were in close agreement with the release profiles suggesting that a diffusion-erosion mechanism was in operation. Considering the hydrophobic nature of the drugs, the release study was carried out maintaining sink conditions by addition of 10% solvent in the release medium. 41, 42 The drug release in drug eluting stents is influence by many factors such as coating morphology, heterogeneous drug distribution, drug nature, degradation rate of polymer, etc.

43

In the present study, the differential

localization of the two drugs in the polymer film may contribute to the difference in the release rates of the two drugs, especially in the initial time points. Atorvastatin is preferentially localized on the surface while fenofibrate partitioned deeper in to the polymer film. This is also reflected in the higher burst release (7%) for atorvastatin when compared to fenofibrate (4.6%). The burst release of

19

ACS Paragon Plus Environment

ACS Biomaterials Science & Engineering

the drugs was observed in first 2-6 h of release, which is followed by a period of sustained release. The burst release is attributed to the drug present in the peripheral regions of the film while the release at the later time points is influenced by the degradation of the polymer chains. The slow release of the drugs at the later time points clearly indicates the surface eroding polymer film. PLCL polymer is known to exhibit slow hydrolytic degradation

44

, which is manifested through the slow

release profiles observed in the present study.

Cumulative release %

[A]

Drug 1 (ATS)

60

[D]

Drug 2 (FF)

50 40 30 20 10 0 0

20

40

60

80

[E]

Time (Days)

[C] 7.8

Drug 1(ATS) 11

Cumulative Release %

[B] Cumulative Releasae %

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 20 of 53

10.95 10.9 10.85 10.8 10.75

Drug 2 (FF)

7.75 7.7 7.65 7.6 7.55

10.7

7.5

10.65

7.45 7.4

10.6 5

7

14

18

24

28

35

42

Time (Days)

50

5

7

14

18

24

28

35

42

50

Time (Days)

Figure 4. [A] In vitro drug release profiles of atorvastatin and fenofibrate from the dual drug-loaded PLCL coated stent. [B] and [C] expanded plots of the release. [D] SEM of dual drug-loaded PLCL coated stent at low and [E] high magnification after 80 days of release.

3.4 In vitro degradation. A gradual change in surface morphology of the dual drug loaded PLCL coating was observed during the degradation study (Figure 5). At initial time points, micro pores were formed in the coating indicating water penetration due to hydrolysis and release of the surface



20

ACS Paragon Plus Environment

Page 21 of 53 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

ACS Biomaterials Science & Engineering

bound drug molecules. At the later time points, the size of the pores was found to progressively increase due to greater permeation of water through the void spaces formed by the release of the surface entrapped drug. The burst release observed in the initial time points correlates with the release surface bound drug. As the non-polar fenofibrate tends to localize in the interior, the burst release and subsequent sustained release due to the surface erosion of the polymer is of lower magnitude for fenofibrate than atorvastatin. These data clearly suggest that a dominant surface erosion of the polymer coating is in operation. Similar observations have been reported by Biggs et al., wherein the relation of drug release from drug-rich region of a commercial drug eluting stent and pore formation on the surface and micro-networks in subsurface area of coating has been described. 43

2nd week [A]

4th week

6th week

8th week

10th week

[B]

Figure 5. SEM images of dual drug loaded stent surface during in vitro degradation study at [A] low and [B] high magnification



21

ACS Paragon Plus Environment

ACS Biomaterials Science & Engineering

3.4 In vitro studies. Figure 6 shows the phase contrast images of the cells in the vicinity of the bare metal and dual drug-loaded stents. Endothelial cells cultured with bare metal and dual drug-loaded PLCL coated stent exhibit normal morphology with no visible effect on their proliferation at each time point (Figure 6). This indicates the biocompatible nature of the dual drug-loaded stent, which can aid re-endothelialization with no adverse effects on wound healing mechanism in the vascular space. In contrast, vascular smooth muscle cells show morphological changes in presence of the dual drug-loaded stent when compared to their morphology in presence of the bare metal stent. Smooth muscle cells surrounding the dual drug-loaded stent tend to form a contracted morphology and do not exhibit an extended shape, indicating a direct response of the cells to the drugs released in to the media (Figure 6). These results demonstrate the cytostatic effect of the combination of atorvastatin and fenofibrate on hVSMC that retard their ability to adhere and proliferate on the dual drug-loaded stent while simultaneously promoting HUVEC growth and migration.

hVSMC

HUVEC Dual drug loaded PLCL stent

Bare metal stent

Dual drug loaded PLCL stent

Day 1

Bare metal stent

Day 3

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 22 of 53

Figure 6. [A] Phase contrast microscopy images of HUVEC and hVSMC cultured with bare metal stent and dual drug eluting stent.



22

ACS Paragon Plus Environment

Page 23 of 53

Figure 7 shows the SEM of the HUVEC and hVSMC on the stent surface. The SEM reveals that no smooth muscle cell is adhered on the surface of dual drug-loaded PLCL coated stents. In contrast, a healthy proliferation of hVSMC on the bare metal stent was observed that progressively covers the stent surface by 72 h. This demonstrates that the dual drug-loaded PLCL coated stent inhibits the hVSMC adhesion and migration, and hence has the potential to curb complications like restenosis. In contrast, the endothelial cell adhesion was observed on the dual drug-loaded stent with an extended morphology suggesting that the dual drug combination favours the adhesion, proliferation and extension of endothelial cells, which is beneficial for endothelialization of the stented site.

HUVEC

hVSMC Dual drug loaded PLCL stent

Bare metal stent

Dual drug loaded PLCL stent

Day 1

Bare metal stent

Day 3

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

ACS Biomaterials Science & Engineering

Figure 7. SEM images of bare metal and dual drug loaded stents after culture with hVSMC and HUVEC for 3 days. Scale bar represents 50 μm and inset at 20 μm.



23

ACS Paragon Plus Environment

ACS Biomaterials Science & Engineering

Figure 8 shows the results of immunostaining assays performed on HUVEC and hVSMC. The immunostaining assay clearly demonstrates the pleiotropic effect of atorvastatin and fenofibrate combination. Alpha smooth muscle actin staining (Figure 8) shows poor cell adhesion and negligible cell-cell communication in hVSMCs after day 1 and day 3 of culture with dual drug-loaded PLCL coated stent when compared to the bare metal stent. In contrast, CD31 (PECAM-1) staining (Figure 8) reveals a well-extended morphology of endothelial cells. The presence of the intercellular junction protein in the cells cultured on dual drug-loaded stent indicates the establishment of a cellular communication network in the endothelial cells. The immunostaining assays confirm that the drugs combination promotes fast endothelialization and lowers the risk of neointimal hyperplasia through retardation of smooth muscle cell adhesion and proliferation. An earlier report has suggested that the atorvastatin and fenofibrate drug combination may exhibit cell specific-effect on the proliferation and migration of smooth muscle cells and endothelial cells in tissue culture polystyrene culture plates.45 Our results confirm this effect of the drug combination on a polymercoated metal stent surface.

hVSMC (α‐SMA) Dual drug loaded PLCL stent

Bare metal stent

Day 1

Bare metal stent

HUVEC ( CD31)

Day 3

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 24 of 53



24

ACS Paragon Plus Environment

Dual drug loaded PLCL stent

Page 25 of 53 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

ACS Biomaterials Science & Engineering

Figure 8. Fluorescence microscopy images of α-SMA and CD31 staining on adhered hVSMC and HUVEC respectively on bare metal and dual drug-loaded PLCL coated stents. Scale bar represents 100 μm.

3.5 Whole blood adhesion and APTT. Figure 9 shows the blood clot formation on the bare metal stent with extensive fibrin network indicating its aggressive thrombogenic nature. This is consistent with the observation reported by earlier groups on stainless steel stents.

46

Comparatively, the

polymer-coated stent shows lesser clot formation. This difference may arise due to the difference in the extent of platelet adhesion on the smooth polymer surface and their activation (Figure 9B). Interestingly, the dual drug-loaded PLCL coated stent displayed no clot formation and had significantly less number of red blood cells adhering to the surface. This result provides a direct evidence of the thrombo-resistant nature of dual drug-loaded PLCL coated stent (Figure 9C). The thrombo-resistance arises due to the pleiotropic effect of the fenofibrate and atorvastatin combination. Atorvastatin has been shown to modulate thrombomodulin levels thereby conferring thrombo-resistance.47 Though no direct evidence of fenofibrate on the thrombus inhibition is available, several studies have demonstrated that fenofibrate improves endothelial function by regulating nitric oxide levels and therefore may contribute to thrombo-resistance indirectly.48 Thus the atorvastatin-fenofibrate combination in PLCL matrix was found to be beneficial in avoiding platelet adhesion, activation and thrombus formation. Figure 9D shows the effect of the dual drugloaded coating on the coagulation time of platelet poor plasma (PPP) using the APTT assay. The results show a significant delay in the clotting time of PPP in presence of dual drug eluting stent when compared to the control indicating that the drug combination exhibits anti-coagulation properties. Few earlier reports have indicated that the drug combination regulates peroxisome proliferative-activated receptors (PPARα)

49

and NADPH oxidase 2 (NOX2), which play role in

platelet activation. 50 The drugs have also been independently and in the combination shown to alter



25

ACS Paragon Plus Environment

ACS Biomaterials Science & Engineering

the levels of coagulation factors, factor VII, fibrinogen and PAI-1.51 Though the exact mechanism responsible for the anti-thrombogenic nature of both the drugs is yet to be deciphered, it is evident that this combination possesses anti-thrombogenic properties through inhibition of platelet adhesion and activation that has been demonstrated in stent coatings for the first time in the present study. [A]

[B]

[C]

[D] 40 APTT (s)

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 26 of 53

*

35 30 25 20 15 10 5 0 Plasma

BMS

PCS

DDES

Figure 9. Scanning electron micrographs of whole blood adhesion on [A] 316S stainless steel stent, [B] polymer coated stent and [C] dual drug-loaded PLCL coated stent. [D] Coagulation times recorded in the APTT test for different samples (*p